TISSUE-SPECIFIC STEM CELLS

Size: px
Start display at page:

Download "TISSUE-SPECIFIC STEM CELLS"

Transcription

1 TISSUE-SPECIFIC STEM CELLS Concise Review: Stem Cells, Myocardial Regeneration, and Methodological Artifacts PIERO ANVERSA, a ANNAROSA LERI, a MARCELLO ROTA, a TORU HOSODA, a CLAUDIA BEARZI, a KONRAD URBANEK, a JAN KAJSTURA, a ROBERTO BOLLI b a Cardiovascular Research Institute, Department of Medicine, New York Medical College, Valhalla, New York, USA; b Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, USA Key Words. Heart Confocal and light microscopy Mitosis Chimerism Bone marrow cell transdifferentiation Enhanced green fluorescent protein autofluorescence ABSTRACT This review discusses the current controversy about the role that endogenous and exogenous progenitor cells have in cardiac homeostasis and myocardial regeneration following injury. Although great enthusiasm was created by the possibility of reconstituting the damaged heart, the opponents of this new concept of cardiac biology have interpreted most of the findings supporting this possibility as the product of technical artifacts. This article challenges this established, static view of cardiac growth and favors the notion that the mammalian heart has the inherent ability to replace its cardiomyocytes through the activation of a pool of resident primitive cells or the administration of hematopoietic stem cells. STEM CELLS 2007;25: INTRODUCTION This article discusses the fundamental results that have contributed to create the view that the postnatal and adult heart cannot renew its parenchymal cells and that the number of myocytes that are present at birth dictates the destiny of this organ throughout life [1, 2]. According to this premise, myocyte turnover does not occur, and the cells originally formed during embryonic and fetal cardiac development are responsible for the preservation of myocardial performance in the young, adult, old, and senescent heart. Coronary vascular endothelial cells (ECs) and smooth muscle cells (SMCs) divide rarely, have a very long cell cycle, and are replaced slowly by division of resident ECs and SMCs within the wall, without changing the interaction between the established, irreplaceable myocytes and the modestly more dynamic coronary vessels. Importantly, circulating progenitor cells or primitive cells mobilized from the bone marrow do not transdifferentiate and therefore are incapable of acquiring the myocyte, EC, and SMC lineages [3]. By necessity, myocyte apoptosis has to be an occasional event of no or, at most, trivial consequence; if not, the heart would rapidly lose a significant number of parenchymal cells, and the small group of myocytes left would be unable to sustain ventricular performance. Together, these concepts project a rather static biological view of cardiac homeostasis and pathology. In addition, this perception of the heart imposes severe limitations on the development of therapeutic strategies that can be implemented for the management of human disease [4]. We will review the work that has promoted a shift in paradigm of the heart from a terminally differentiated postmitotic organ to a self-renewing organ. This dramatic change in understanding of cardiac behavior and function was originated by observations that were made in the 1940s and that have continued to accumulate over the years (for reviews, see [5, 6]). However, the disagreement that persists among the members of the scientific community today was present then and has persisted for nearly 60 years. The reason for the controversy is unclear, but it seems to reflect unshakable positions based on preconceived beliefs more than on careful analysis and proper consideration of published results [7]. In this article, we focus on methodological issues because, in the course of the controversy, recurrent statements have been made to undermine the technical protocols used in studies supporting the existence of cardiac regeneration. Consistently, the data in favor of myocardial regeneration are claimed to be the product of methodological artifacts [1 4, 8 11]. To properly evaluate the validity of these criticisms or lack thereof, methodologies need to be compared to provide readers with a better understanding of the substrate that governs the debate. The old dogma has profoundly conditioned basic and clinical research in cardiology for the last three decades [7, 12]. Based on this paradigm, cardiomyocytes undergo cellular hypertrophy but cannot be replaced either by the entry into the cell cycle of a subpopulation of nonterminally differentiated myocytes or by the activation of a pool of primitive cells that become committed to the myocyte lineage. However, the efforts made to introduce a highly dynamic perspective of the heart have led to the identification and characterization of a resident pool of stem cells that can generate myocyte, and ECs and SMCs organized in coronary vessels [13]. This discovery has created a new, heated debate concerning the implementation of adult cardiac stem cells in the treatment of heart failure of ischemic and nonischemic origin. Correspondence: Piero Anversa, M.D., Cardiovascular Research Institute, Vosburgh Pavilion, New York Medical College, Valhalla, New York 10595, USA. Telephone: ; Fax: , piero_anversa@nymc.edu Received October 3, 2006; accepted for publication November 7, 2006; first published online in STEM CELLS EXPRESS November 22, 2006; available online without subscription through the open access option. AlphaMed Press /2007/$30.00/0 doi: /stemcells STEM CELLS 2007;25:

2 590 Controversy in Cardiac Repair POSSIBILITY 1: THE HEART IS A TERMINALLY DIFFERENTIATED POSTMITOTIC ORGAN The majority of organs contain dividing and nondividing cells. However, nondividing cells can rest in the G 0 phase and reenter the cell cycle following growth activation or become terminally differentiated and die without undergoing further division [14]. The latter category of cells is not dormant in G 0 and cannot reenter the cell cycle. This is typically seen at the tip of the villi of the small intestine [15], in the auditory hair cells of the ear [16], and in the upper layer of the epidermis [17]. Hepatocytes in vivo are in a G 0 state, and after partial hepatectomy or severe injury, liver regeneration is accomplished by proliferation of hepatocytes, biliary epithelial cells, and fenestrated endothelial cells [18]. Because the liver contains a population of primitive cells, the oval cells, the growth and maturation of this cell category may contribute to the reconstitution of liver mass [19]. Probably both cell mechanisms are involved in the repair of the organ, although controversy exists in the field [20]. Multipotential cells have also been identified in the central nervous system, but questions have been raised concerning their ability to grow and generate Purkinje neurons in the adult brain [21 24]. Conversely, the regeneration of the skin [25], bone marrow [26], and skeletal muscle [27] is regulated by activation of skin stem cells, hematopoietic stem cells (HSCs), and satellite cells, respectively. These mechanisms of cellular growth have not been considered possible in the myocardium, and the notion has been proposed that ventricular myocytes cannot be replaced once cell division ceases immediately after birth in the mammalian heart [1, 2, 9, 10]. Thus, the dogma was established that the postnatal heart is composed of a fixed number of myocytes and that, if myocytes die, they are permanently lost and the myocardium must maintain its vital role with a reduced number of cells. The remaining myocytes are not in G 0 and cannot be triggered into the replicating phase [28]; they continue to perform their physiological function, undergo cellular hypertrophy, and ultimately die [29]. Based on this paradigm, the age of myocytes, organ, and organism was assumed to coincide, implying that myocytes in humans may have a lifespan that exceeds 100 years [30]. For several decades, no effort was made to reexamine this rather unusual view of the biology of the heart and cardiac homeostasis. Remarkably, there is not a single piece of evidence that demonstrates the inability of the heart to replace its dying myocytes. It seems rather extravagant that cardiomyocytes can contract 70 times per minute over 100 years and continue to be functional. During this period, they would have contracted 3.7 billion times and still be operative. If this were to be the case, adult myocytes would be essentially immortal cells. POSSIBILITY 2: THE HEART IS NOT A TERMINALLY DIFFERENTIATED POSTMITOTIC ORGAN Historically, the foundations for the view of the heart as a terminally differentiated postmitotic organ incapable of regeneration were established in the mid-1920s [31]. This conclusion was dictated by the difficulty in identifying mitotic figures in myocyte nuclei of the human heart with the unsophisticated histologic preparations available years ago. This notion gained support from autoradiographic studies of tritiated thymidine incorporation in the myocardium during postnatal development and pathological overloads [32 35], conducted in the mid- and late 1960s. DNA synthesis in myocyte nuclei was negligible, and this observation, together with the inability to recognize myocyte mitotic figures, led to the belief that the adult heart is composed of parenchymal cells that are permanently withdrawn from the cell cycle. Myocytes can increase in volume but not in number [36]. Again, this work used standard histologic preparations in combination with photographic emulsions, which imposed severe limitations in terms of microscopic resolution [37]. These factors made it highly problematic to distinguish whether DNA replication or mitosis was present exclusively in interstitial cells or, at times, involved cardiomyocytes as well. In fact, the possibility of myocyte division was acknowledged in two of the early reports [34, 35]. The qualitative results discussed so far were in sharp contrast with quantitative measurements of myocyte volume and number performed in human hearts obtained from patients who died because of decompensated cardiac hypertrophy and congestive heart failure [5]. In the late 1940s and early 1950s, Linzbach documented that, in the presence of a cardiac weight equal to or greater than 500 g, myocyte proliferation represented the predominant mechanism of increased muscle mass [38, 39]. An interesting aspect of this work is that these determinations were all based on two very simple but critical parameters: myocyte length, assessed by the distance between two nuclei of largely mononucleated myocytes, and myocyte diameter across the nucleus. These variables are easily recognizable in standard histologic sections and do not require high resolution and an impeccable preparation. Based on a cylindrical shape configuration, myocyte cell volume was computed. Moreover, the aggregate volume of the myocyte compartment of the myocardium was obtained, and the quotient of this quantity and myocyte cell volume yielded the total number of ventricular myocytes. Because of the simplicity of the approach, the original results were confirmed several years later in other laboratories in which more refined techniques were applied [40, 41]. In all cases, hearts weighing 500 g or more were characterized by a striking increase in myocyte number that was more prominent than cellular hypertrophy; this adaptation involved both the left and right ventricles [38 43]. In the mid- and late 1990s, new studies of the human heart examined the distribution of mononucleated and binucleated myocytes in 72 normal and 176 diseased hearts [43]. Aging, cardiac hypertrophy, and ischemic cardiomyopathy were characterized by the lack of changes in the relative proportion of mononucleated and multinucleated myocytes in the ventricular myocardium, confirming that the early and more recent measurements of myocyte proliferation were valid and did not represent and erroneous interpretation dictated by nuclear hyperplasia in the absence of myocyte division [6, 44]. Mononucleated cells constitute 75% of myocytes in the human heart, differing significantly from mice [45], rats [46], dogs [47, 48], and pigs [49]. Mononucleated cells are smaller than binucleated cells, and this cellular property may influence the ability of myocyte to divide [50]. Human myocytes larger than 30,000 m 3 cannot reenter the cell cycle and proliferate [50]. In fact, cycling and mitotic myocytes are predominantly small and mononucleated, and this feature may account for the massive cardiac hypertrophy that can be achieved in humans (Fig. 1). It would be inefficient for large myocytes to divide once or at most twice to expand the cardiac mass. Heart weight in humans can increase nearly threefold, reaching values of 1,000 g or larger [5, 38, 41, 43]. Heart failure typically shows increases in myocyte number that vary from 20% 100% or more [5, 38, 41, 43, 51 53]. This phenomenon is not affected by age; in an analysis of 7,112 human hearts, from birth to 110 years of age, Linzbach and

3 Anversa, Leri, Rota et al. 591 Figure 1. Myocyte proliferation and ischemic cardiomyopathy in humans. Small dividing left ventricular myocytes ( -sarcomeric actin; red) are identified by Ki67 labeling (A) (white, arrow), MCM5 expression (B) (yellow, arrow), and metaphase chromosomes (C) (arrow). The boundaries of myocytes and interstitial cells are defined by laminin (green). Nuclei are stained by 4,6-diamidino-2-phenylindole (blue). Akuamoa-Boateng have shown that extreme forms of organ hypertrophy are detectable up to the ninth decade of life, and heart weights of 500 and 600 g are present in patients at 100 years of age and older [54]. Importantly, scar formation, foci of replacement fibrosis scattered throughout the ventricular wall, diffuse interstitial fibrosis, and ongoing myocyte death by apoptosis and necrosis are typically present in the hypertrophied failing heart [44, 55, 56]. These factors markedly affect the number of cardiomyocytes and indicate that the extent of myocyte formation determined by the morphometric methodologies summarized above constitutes an underestimation of the actual magnitude of myocyte regeneration in the pathological heart. In addition, there is no alternative methodology able to demonstrate unequivocally myocyte proliferation. By definition, myocyte replication corresponds to an increase in the number of myocytes; in the absence of this documentation, biochemical, molecular, physiological, autoradiographic, and structural results can only be suggestive of myocyte regeneration but in themselves cannot provide the critical information, that is, an increased number of cells. Quantitative morphology is the only technique that can identify myocyte formation. Contrary to expectations, the observations in favor of myocyte regeneration were not received with enthusiasm by the scientific community [57 63]. With some small variations, the morphometric results in the human heart were questioned, and the possibility of myocyte formation in the adult myocardium was first dismissed and subsequently attacked. This new idea challenged the comfortable paradigm of myocyte hypertrophy that had dominated the field of molecular cardiology and cell signaling for the previous 25 years. Heart failure was viewed as the consequence of a defective myocyte hypertrophy in which alterations of effector pathways regulating myocyte growth and contractility were considered responsible for the depression in organ, tissue, and cell function [64, 65]. However, the quantitative data collected in the human heart suggested that a reexamination of the mechanisms of myocyte growth needed to be considered and that additional studies needed to be performed to clarify the apparently contradictory results. For this purpose, experiments were conducted in animal models of cardiac failure and in human diseased hearts to provide relevant information in support of or against the notion of the regeneration potential of the adult myocardium. New methodologies were introduced to determine whether a subset of cardiomyocytes had the ability to reenter the cell cycle and divide and whether this process involved the activation of cell cycle-related genes, cyclins, and cyclin-dependent kinases and the expression of proteins modulating karyokinesis and cytokinesis [45, 50, 55, 66 80]. Rounds of cell division result in progressive telomere attrition, and critical telomeric shortening triggers irreversible growth arrest [81 83]. Thus, the telomeretelomerase axis, together with the telomere-binding proteins, is a fundamental component of the multiple mechanisms that regulate the regeneration of non-postmitotic organs; because of this function, this system was evaluated and found to be operative in the adult heart of animals and humans [50, 74 77, 79, 84]. Unfortunately, these multiple studies left the controversy unresolved. MYOCYTE DIVISION In 1925, a highly significant paper was published in which the claim was made that there are no detectable mitotic figures in myocytes of the human heart, and therefore myocyte regeneration does not occur in mammals [31]. This work challenged the view proposed in numerous studies published from 1850 to 1911 in which the general belief was that cardiac hypertrophy is the consequence of hyperplasia and hypertrophy of existing myocytes [6]. The 1925 study can be considered the one that introduced the concept that the heart is a terminally differentiated postmitotic organ. The impact of this research was enormous. To the best of our knowledge, for several decades, many generations of pathologists and cardiovascular scientists have accepted the view that mitotic myocytes are not to be found in the adult myocardium, and only a few rare exceptions have been published, mostly restricted to the fetal-neonatal heart [85 88]. In the last years, however, mitotic images in human cardiomyocytes have been documented in acute and chronic ischemic cardiomyopathy [72, 89, 90], idiopathic dilated cardiomyopathy [72], chronic aortic stenosis with moderate ventricular dysfunction [50], myocardial aging [84], acromegaly (Fig. 2A), and diabetes (Fig. 2B). Using light microscopic examination of routine histologic sections, an extensive search was required for the recognition of occasional mitotic figures in myocytes. Because in the early successful observations, dividing myocytes were seen only in the fetal and diseased failing heart [89], the assumption was made that replicating myocytes are essentially undetectable in normal adult myocardium. This hypothesis, in fact, suggested a rather minor role of myocyte regeneration in cardiac homeostasis and pathology in humans. However, in spite of the caution exercised in the interpretation of these results, this work was considered to reflect the incorrect interpretation of proliferating interstitial fibroblasts as dividing myocytes [1, 2]. Surprisingly, this conclusion was reached based on electron microscopic images that illustrated nondividing fibroblasts in proximity to differentiated nonreplicating cardiomyocytes [1]. This represents the normal organization of the myocardium and makes it difficult to understand how this well-established morphological characteristic of cardiac structure can be used as an argument against myocyte division and a case in favor of its erroneous identification. So Artifact 1 was introduced in the literature as the only logical explanation for these unexpected results. An important point that necessitates discussion is the belief by some that electron microscopy is the only tool that can unequivocally prove or disprove the presence of myocyte mitotic images in the myocardium. In invited reviews questioning the existence of myocyte regeneration, interstitial fibroblasts have been described as extremely mobile cells capable of penetrating by several microns the thickness of myocytes, positioning themselves in the central portion of

4 592 Controversy in Cardiac Repair Figure 2. Myocyte generation in the diseased human heart. Metaphase chromosomes (blue, 4,6-diamidino-2-phenylindole [DAPI]; arrows) are apparent in two small dividing cardiomyocytes (red, -sarcomeric actin) in patients affected by acromegaly (A) or diabetes (B). The organization of tubulin in the mitotic spindle (C) (white, arrows), the accumulation of actin in the contractile ring (D) (red, arrows), and cytokinesis (E) (arrows) in dividing cardiomyocytes are apparent. In all cases, chromosomes are labeled by DAPI (blue) and the myocyte cytoplasm by -sarcomeric actin (red). Green indicates laminin. these cells [1, 2]. By this mechanism, the myocyte nucleus was apparently displaced from its original location, which was occupied by the penetrating fibroblast. Schemes were created to strengthen this unlikely possibility [1]. However, this highly imaginative understanding of cardiac morphology fell short on several accounts: the examples of penetrating interstitial cells did not show dividing cells, the myocyte nucleus was at its expected site in the middle of the myocyte cytoplasm, and the electron microscopic preparation was of poor quality. Myocytes were hypercontracted and had sarcomeres of m in length. Normally, diastolic sarcomere length is m, and during systolic contraction, it becomes m [91]. Hearts with sarcomeres 1.4 m in length are dead, since no contraction can be generated under this condition. Contraction bands produced by the inappropriate fixation of the myocardium were analyzed to reinterpret myocardial ultrastructure. Surprisingly, these artifacts were accepted and introduced in the literature as facts. The frequency of mitosis in myocytes of normal human, mouse, or rat heart, as assessed by confocal microscopy, is low and averages 14/10 6 myocytes [72], 37/10 6 myocytes [45], and 85/10 6 myocytes (J.K., unpublished data), respectively. The values for dividing interstitial fibroblasts are similar to those of cardiomyocytes [89]. Based on these data, 350, 140, and 60 mm 2 of tissue need to be examined to identify a dividing myocyte in the human, mouse, and rat heart, respectively. This is why the suggestion to implement electron microscopy for this analysis is not realistic. The area of myocardium that is sampled in each thin section by electron microscopy is 0.2 mm 2, and the area of the microscopic field at a magnification of approximately 2,000 is mm 2. Thus, the numbers of micrographs that would have to be collected to identify a single myocyte mitotic image are 66,000 pictures for humans, 25,000 for mice, and 11,000 for rats. If one wishes to use electron microscopy to challenge the existence of myocyte division and to demonstrate that proliferating fibroblasts were improperly interpreted as mitotic myocytes, that is what needs to be invested to obtain at least one example in support of or against this thesis [92]. Major advances in understanding myocyte replication in the adult human heart have been made by the use of confocal microscopy and immunolabeling in the analysis of the normal and diseased heart. With this approach, the identification of mitotic images in myocytes became easier and more accurate. These new results strengthened the notion that parenchymal cells are formed continuously in the normal myocardium and myocyte regeneration is markedly potentiated in the pathological failing heart [6, 50, 72, 84, 90]. This technology was also applied to the study of animal models of human diseases, and myocyte replication was unequivocally demonstrated in myocardial sections and isolated cell preparations [6, 45, 73, 93]. The latter observation conclusively refuted the claim that dividing myocytes represent proliferating fibroblasts inserted within or attached to terminally differentiated myocytes. The formation of the mitotic spindle by the arrangement of tubulin and the generation of the contractile ring by the accumulation of actin in the region of cytoplasmic separation were clearly documented, together with karyokinesis and cytokinesis (Fig. 2C 2E). However, these results did not change the position of certain groups, who continued to reject the regenerative ability of the adult mammalian myocardium [2, 4, 8, 9, 28]. These groups introduced the possibility of a new source of artifacts, Artifact 2, related to confocal microscopy; they stated that traditional light microscopy is greatly superior to confocal microscopy and, with this personal viewpoint, tried to invalidate the results obtained with the latter technique. An important issue that requires discussion relates to the notion that the analysis of histochemical preparations by conventional light microscopy is superior to fluorescence labeling and confocal microscopy [94, 95]. The resolution of the micrographs provided by light microscopy is markedly inferior to that obtained by confocal microscopy, 0.62 versus 0.29 m [37, 96]. In addition, only the very superficial layer of a tissue section can be examined by light microscopy (Fig. 3). This inherent problem with light microscopy was recognized immediately when confocal technology became available [97]. Clear examples were published in the Journal of Cell Biology in 1987 demonstrating the impossibility of identifying microtubules, mitotic spindle, cytoplasmic proteins, and chromosomal structures by light microscopy in cultured cells. Conversely, these morphological details were apparent when the same cells were examined by confocal microscopy [98]. The difference between epifluorescence light microscopy and confocal microscopy is even greater in tissue sections. When confocal microscopy was not available to us, we estimated the myocyte mitotic index in chronic heart failure to be 11 myocytes per million [89]. When comparable hearts were evaluated by confocal microscopy, the mitotic index reached a value of 152 cells per million [72]. Even more striking is the difference between the mitotic indices obtained by light and confocal microscopy in the border zone of acute infarcts in humans: light microscopy yielded 3.3 mitotic myocytes per million [89] and confocal microscopy 775 mitotic cells per million [90]. In all cases,

5 Anversa, Leri, Rota et al. 593 Figure 3. Light epifluorescence microscopy of human myocardial tissue sections. Panels show different focal planes. (A): Superficial layer of the section. (B): Focal plane located 1 m below the surface. (C): Focal plane located 2 m below the surface. Note that sarcomere striations (green, -sarcomeric actin) and details of nuclear structure (red, propidium iodide) are well-defined only in (A) and are not clearly visible in (B) and (C). The yellowish area adjacent to the nucleus reflects the accumulation of lipofuscin. the measurements of myocytes in mitosis were made in the same laboratory. CARDIAC CHIMERISM AND MYOCYTE FORMATION The controversy concerning the recognition of dividing myocytes in the adult heart intensified when studies on cardiac chimerism were conducted in an attempt to determine whether myocardial regeneration could be accounted for by the activation of undifferentiated cells of recipient origin [99]. Although studies of the chimerism of the heart and other organs following sex-mismatched cardiac or bone marrow transplantation have provided consistent results concerning the migration of primitive cells from the host to the graft [94, 95, ], findings from different laboratories have been inconsistent in terms of the extent of this process, and this variability has promoted a heated debate in the scientific community. After homing, host progenitor cells undergo replication and differentiation generating mature parenchymal cells and vascular structures in the transplanted organ. There is little disagreement among authors on the occurrence of this phenomenon in sex-mismatched heart transplant, but the magnitude of cardiac chimerism varies significantly in different reports [94, 95, , 104, 109, 110]. This discrepancy involves mostly ventricular myocytes and, to a much lesser extent, newly formed coronary vessels. For myocytes, the published values range from as high as 18% to as low as 0.02% or absent [99, 100, 104]. A plethora of intermediate results has also been obtained. The surprising outcome of these findings was that the low extent of myocyte chimerism detected by some groups was used to question myocyte formation and to support the traditional view that the heart is a terminally differentiated postmitotic organ incapable of undergoing meaningful regeneration of any kind [2, 4, 8 10]. However, to accomplish this task, a rational explanation was necessary to account for the data pointing to a high number of male myocytes within female hearts transplanted in male recipients [99]. In fact, these observations indicated that a significant proportion of male myocytes was present in the female heart as a result of activation and commitment of the host male progenitor cells. Interestingly, the discrepancy in the degree of cardiac chimerism was immediately adopted to initiate another controversy and introduced in the literature Artifact 3. The promoters of this new source of technical error reached this conclusion by assuming that only the absence or minimal levels of chimerism were correct, whereas high levels of chimerism had to be necessarily wrong [94, 95, 104, 115]. The critics decided to take the prerogative of reinterpreting data published by other groups, pointing to a number of crass morphological errors. Surprisingly, these negative comments were published and, once again, these questionable criticisms became facts. The most obvious technical differences among the studies published so far is the use of conventional light microscopy [94, 95, , 104] versus confocal microscopy [99, 109, 110]. The probes used for the detection of the Y chromosome in female hearts transplanted in male recipients were not always the same, and the procedures used for the recognition of the Y chromosome differed as well. These factors introduced critical variables in the acquisition of the data that, together with the method of analysis, have been a major cause for the discrepancy. A serious technical and conceptual concern is that similar high values of Y-chromosome-positive myocyte nuclei have been obtained in control male human hearts, averaging 50%. However, the values reported in control male human hearts by conventional fluorescence microscopy and confocal microscopy in the absence of optical sectioning of the samples are unrealistic and therefore cast doubts on this series of studies. By these two methods of analysis, the fraction of Y-chromosome-positive myocyte nuclei is low and rarely exceeds 15% (Fig. 4A). This is due to the limited focal depth of the objective, 0.5 m, [37] and the extent by which a nucleus has to be embedded within the section to be visible (i.e., the penetration factor) [44]. Moreover, with conventional fluorescence microscopy, the out-of-focus fluorescence blurs the image, complicating the identification of the Y-chromosome signal in nuclei (Fig. 4B). When histologic sections of male myocardium are examined by confocal microscopy, the superficial layer of the section, which corresponds to 0.6 m, shows 12% Y-chromosome-positive myocyte nuclei (Fig. 4C). Importantly, the evaluation of the entire thickness of the section by the analysis of consecutive optical planes markedly increases the detection of Y-chromosome-labeled myocyte nuclei, reaching a cumulative value of 55% (Fig. 4D). Therefore, in several reports in which conventional fluorescence microscopy or confocal microscopy restricted to a single plane was used [94, 101, 102, 104], the finding of the Y chromosome in a large number of myocyte nuclei of control male hearts is at variance with reality. Because of this unacceptable premise, the extremely low number or absence of Y-chromosome-positive myocyte nuclei found in the transplanted female hearts can be seriously questioned. The volume of a myocyte nucleus is 250 m 3, and the signal for the Y chromosome has a volume of 0.5 m 3, occupying 0.2% of the entire nucleus. When the Y chromosome is present in a mid-section of a nucleus, the area of the Y- chromosome signal is 1 m 2, and the area of the nucleus profile is 75 m 2 [116]. Thus, the Y chromosome occupies 1.3% of the nuclear area (Fig. 4E). It follows that the probability of hitting the Y chromosome in myocyte nuclear profiles of a tissue section can be calculated. Importantly, the chance of a positive signal varies as a function of section thickness and is influenced by the orientation of the nuclei in myocytes. These variables can be computed according to the following equations [44, 116].

6 594 Controversy in Cardiac Repair Figure 4. Detection of Y-chr. by fluorescence in situ hybridization. (A): One myocyte nucleus positive for the Y-chr. (white dot, arrow) is visible by conventional epifluorescence microscopy of 6- m-thick section of human male myocardium. Seven nuclei pertaining to myocytes ( -sarcomeric actin, red, arrowheads) are present in the field. (B): Often, out-of-focus fluorescence makes it difficult the recognition of Y-chr. labeling. The arrowhead points to a nuclear dot that may correspond to the Y-chr. signal. (C, D): Confocal microscopy of a 6- m-thick tissue section of human male myocardium; the two panels illustrate the same field. (C): Superficial layer of the section. (D): Combination of 10 optical projections of 0.6 m each of the entire section thickness. Arrowheads indicate Y-chr.-positive myocyte nuclei in (C) (2 of 15) and (D) (9 of 16). Laminin (green) defines the periphery of the cells. (E): High-magnification confocal image of a myocyte nucleus positive for the Y-chr. The area occupied by the nucleus is 176 m 2, whereas the area occupied by the Y-chr. is 2.0 m 2, accounting for 1.1% of the nuclear cross-sectional area. Abbreviation: Y-chr., Y chromosome. P d Y /l N ) F F 1 k/ k t S )) k [w N sin( )] [l N cos( )] In these equations, P corresponds to the probability of finding a Y-chromosome signal with a diameter d Y within a nucleus whose length is described by l N and width by w N. The parameter F represents the fraction of the nucleus embedded in the section. F is a function of the thickness of the section, t S, and the orientation and size of the nucleus, described by the variable k. The angle between the plane of the section and the long axis of the nucleus is given by. Supplemental online Table 1 lists the predicted minimal, maximal, and average values of Y-chromosome-positive myocyte nuclei in control male human hearts according to section thickness, nuclear orientation, and modality of analysis. Surprisingly, there is a dramatic inconsistency between the expected values of Y-chromosome labeling of myocyte nuclei in normal conditions and in published results. Because of the questionable validity of control values in these studies, the number of male cells identified in female-transplanted hearts suffers from lack of reliability and necessitates reevaluation. This issue is certainly more relevant than the claim concerning the difficulty of distinguishing myocyte nuclei from interstitial cell nuclei [1, 2, 94, 115]. As recently emphasized, an experienced pathologist can easily differentiate an interstitial cell from a cardiomyocyte or discriminate an interstitial cell nucleus from a myocyte nucleus, particularly by confocal microscopy [117]. This is confirmed by the clear recognition of the Y-chromosome signal and nuclear profiles by confocal microscopy [99, 109, 110] and the rather ill-defined Y-chromosome labeling and nuclear boundary by light microscopy [94, 95, 104]. In some cases, by light microscopy, the Y chromosome cannot be distinguished from the nucleolus [94] or properly identified because of little contrast, weak intensity of the signal, and diffuse pattern of staining [104]. Several other factors have to be considered when human myocardium is examined. The age of the patients, time from transplantation, level of rejection, immunosuppressive regimen, and, most importantly, the condition of the tissue to be studied may all contribute to the variability in the levels of cardiac chimerism reported in the last several years. Poor fixation and/or delayed fixation affect the integrity of the DNA and, thereby, the probability of detecting any nucleotide sequence [118]. DNA degradation precedes the alterations in protein antigenicity [119] so that Y-chromosome labeling of myocyte nuclei may constitute a significant underestimation of the actual degree of myocyte chimerism present in the organ [109, 117]. Likewise, prolonged preservation of the myocardium in formalin fixative produces excessive cross-linking of proteins [120], and this condition diminishes the accessibility of the Y-chromosome probe to the nuclear DNA [118, 121]. It is surprising that these crucial factors, together with elementary morphometric principles, have been ignored in the studies pointing to the absence or minimal levels of cardiac chimerism following sex-mismatched heart allograft or bone marrow transplantation. An elegant protocol has been introduced in one study of cardiac chimerism following sex-mismatched bone marrow transplantation to correct, at least in part, this problem. The fluorescence in situ hybridization (FISH) assay was performed with two distinct probes, which detected, respectively, the Y chromosome and the X chromosome in myocyte nuclei [103, 109]. Nuclei negative for the X chromosome had severely altered DNA and had to be excluded from the analysis. When this approach was used, the extent of cardiomyocyte chimerism was found to be 30-fold higher than in other comparable studies [108]. Because questions have been raised concerning some of the values obtained in our study of cardiac chimerism following sex-mismatched heart transplantation [99], we have re-examined cases with the highest levels of Y-chromosome-positive myocyte nuclei using this dual-color FISH assay and evaluated the percentage of XX-positive and XY-positive myocyte nuclei (Fig. 5A, 5B). The original data were confirmed and strengthened by this more sophisticated approach. In addition, the measurements of the number of X and Y chromosomes in nuclei allowed us to exclude cell fusion as the mechanism of male myocyte formation in the female heart. Importantly, optical sectioning by confocal microscopy prevented any possible misinterpretation of interstitial cell nuclei as myocyte nuclei (Fig. 5C 5L). Together, the studies on cardiac chimerism in patients who had sex-mismatched bone marrow transplantation provide strong evidence that circulating progenitor cells mobilized from the bone marrow have the ability to repopulate the heart with cardiomyocytes and coronary vessels [103, 108, 109]. The generation of parenchymal cells and vascular structures mediated by engraftment and differentiation of primitive cells from the recipient is even greater following heart transplantation [99, 109]. However, in this case, the source of

7 Anversa, Leri, Rota et al. 595 Figure 5. Cardiac chimerism in humans. Confocal images of a tissue section obtained from a female heart transplanted in a male recipient. (A, B): Nuclei (4,6-diamidino-2-phenylindole, blue) are positive for the X chromosome (magenta dots) and Y chromosome (white dots, arrowheads). The coexpression of X and Y chromosomes in the same nucleus indicates male diploid genotype. The majority of nuclei belong to myocytes (B) ( -sarcomeric actin, red). (B): Open arrow points to a male myocyte. The periphery of the cells is defined by laminin (green). (C L): Y chromosome (white dots) and X chromosome (magenta dots) are detected in nine distinct optical sections 1 m apart (D L) of the area indicated by the inset in (C). The Y chromosome in the myocyte nucleus (C) (arrowhead) is not visible in (D) and (E) but is evident in (F H). It is no longer present in (G L). The X chromosome in the same myocyte nucleus is apparent only in (J) and (K). The nucleus of an interstitial cell is also visible in (C) (asterisk). the cells that translocate from the host to the implanted heart remains uncertain [99]. The progenitor cells could have been of hematopoietic origin and migrated to the heart through the systemic circulation, or they could have come from the cardiac remnant preserved during surgery for the attachment of the donor heart [122]. These possibilities are not mutually exclusive. The recognition that cardiac chimerism occurs rapidly and in a noticeable manner has had a dramatic impact on our understanding of the biology of the heart and mechanisms of myocardial repair. This work has formed the foundation for the concept of myocardial regeneration and has created the opportunity for testing the role that HSCs have in the restoration of the injured heart [123]. The notion that HSCs retain a significant degree of developmental plasticity and can acquire cell lineages distinct from the organ of origin has promoted the entire field of cell therapy of the failing heart [124]. Opponents of this emerging new paradigm in cardiology, however, continued to claim that these concepts are the product of artifacts [1, 2, 10, 94, 95, 108, 125]. A recurrent unqualified statement is that they have introduced in their work rigorous and stringent criteria, which implies that others do not adhere to the same high scientific standards. The old paradigm has to survive, and the notion that myocytes cannot be generated postnatally, in adulthood or senescence, must be defended. An example of this approach can be found in a recent report in which levels of EC chimerism of 25% and vascular SMC chimerism of 3.5% have been found

8 596 Controversy in Cardiac Repair Figure 6. Bone marrow cell transdifferentiation. Male c-kit-positive bone marrow cells were obtained from EGFP-transgenic mice and injected shortly after infarction in the female mouse heart. (A C): The regenerated myocardium is composed of small myocytes positive for -actinin (A, C) (red) that express EGFP (B, C) (green) and have a male genotype (C) (Y chromosome, white dots). (D F): Again, the new myocytes express -actinin (D, F) (red) and EGFP (E, F) (green). The vast majority of myocyte nuclei are positive for the cell cycle marker BrdU (F) (white). (G I): The regenerated myocytes express -actinin (G, I) (red) and EGFP (H, I) (green), and the nuclei are positive for MEF2C (I) (yellow). (J L): A newly formed arteriole (J, L) ( -smooth muscle actin, red) expresses EGFP (K, L) (green); smooth muscle cells are positive for GATA-6 (L) (white). (M O): The formed capillaries (M, O) (vwf, red) are EGFP-positive (N, O) (green), and endothelial cells express the transcription factor Ets1 (O) (white). Abbreviations: BrdU, 5-bromo-2 -deoxyuridine; EGFP, enhanced green fluorescence protein; vwf, von Willebrand factor. in sex-mismatched heart transplants in the absence (0.0016%) of myocyte chimerism [95]. HEMATOPOIETIC STEM CELLS AND MYOCYTE FORMATION In the last decade, major discoveries have been made concerning the biology of adult HSCs. They can differentiate into cell lineages distinct from the organ in which they reside and into cells derived from a different germ layer [126]. These properties were considered to be restricted to embryonic stem cells, and because of their dramatic biological and clinical implications, they triggered a vigorous debate in the scientific community. The controversy reached unexpectedly high levels of intensity when HSCs were shown to be able to migrate to sites of injury repairing damage in various organs, in particular the brain and the heart [ ]. The brain and the heart have always been viewed as nonpermissive organs for exogenous and endogenous tissue regeneration. Studies presenting positive results [127, ] were immediately confronted by negative reports [ ], which challenged stem cell plasticity and the emerging new paradigm of organ and organism homeostasis and repair [ ]. Although the possibility that HSCs differentiate into Purkinje neurons was questioned based on data interpretation or inability to reproduce similar findings, a different reaction occurred in response to the documentation of the efficacy of adult HSCs in myocardial regeneration after infarction. Several laboratories have used genetically labeled HSCs from transgenic animals to promote the restoration of infarcted myocardium experimentally to identify the progeny formed [129, ]. HSCs expressing enhanced green fluorescence protein (EGFP) and sex-mismatched transplantation have been used frequently to have two independent markers for the recognition of newly generated myocardium: EGFP expression and Y-chromosome labeling [129, 134, 135]. 5-Bromo-2 -deoxyuridine (BrdU) was also administered chronically to animals after infarction and cell treatment to have a third marker of cell accumulation within the infarcted region (Fig. 6A 6F). Following the detection of EGFP-, Y-chromosome-, and BrdU-positive cells, transcription factors and cytoplasmic proteins specific to myocytes and vascular smooth muscle and endothelial cells were demonstrated (Fig. 6G 6O) so as to avoid potential questions on the validity and accuracy of the results [129, 135]. However, these observations were attacked on methodological grounds and were claimed to be the product of a new

9 Anversa, Leri, Rota et al. 597 Figure 7. Autofluorescence of tissue sections and detection of EGFP in skeletal muscle. Autofluorescence (green) of tissue sections obtained from skeletal muscle after fixation with paraformaldehyde and glutaraldehyde (A) or formalin (B), both in the absence of immunolabeling. Nuclei are stained with 4,6-diamidino-2-phenylindole (blue). (C, D): After fixation with paraformaldehyde and glutaraldehyde, skeletal muscle tissue sections were labeled with -skeletal actin (C) and EGFP (D) antibodies. The lack of sarcomere striation indicates that only unspecific labeling of these two antigens was obtained with this protocol. The signals in (C) and (D) correspond to autofluorescence. In this regard, compare (D) with (A). After fixation with formalin only, skeletal muscle tissue sections were labeled with -skeletal actin (E, G) (red) and EGFP (F, G) (green) antibodies. The presence of striations in the cell cytoplasm indicates that specific labeling was obtained by this approach. Note the coexistence of EGFP-positive and EGFP-negative fibers in the same muscle. (H J): After fixation with formalin, skeletal muscle tissue sections were labeled with -skeletal actin (H, J) (red) and EGFP (I, J) (green) antibodies conjugated with quantum dots. The presence of striations in the cell cytoplasm indicates the specificity of labeling. With this approach, the autofluorescence of a tissue section can be totally avoided: the confocal image illustrated in (K) was acquired with the excitation and emission wavelengths used for quantum dot 655. Note the complete absence of autofluorescence. Importantly, the excitation and emission wavelengths used for quantum dot analysis are distinct from the autofluorescence wavelength of tissue sections. Abbreviation: EGFP, enhanced green fluorescence protein. artifact [2, 9, 10, 138, 139, 144]. The characteristics of Artifact 4 were unexpected because a new twist was introduced in the erroneous interpretation of morphological images obtained by immunolabeling and confocal microscopy: autofluorescence. How these multiple markers of myocardial reconstitution by HSCs could all be the result of autofluorescence is an interesting issue, which unfortunately was never addressed. The autofluorescence was introduced by the same authors who raised the issues of Artifacts 1 3 and who, this time, joined forces with experimental hematologists opposing the concept of HSC plasticity [8]. So the never-documented autofluorescence artifact was invoked as the only logical explanation for the possibility that cardiac repair can be accomplished by exogenous cells. Criticisms have focused on the need to use genetic markers to demonstrate whether HSCs can form heart muscle [2 4, 9, 10]. Genetic markers have been used in the questioned positive studies [129, 135], as well as in the negative reports that have been claimed to be valid [ ]. Both positive and negative publications used immunostaining, but in the former case, autofluorescence was assumed to be a factor, whereas in the latter case, the technique used was considered immaculate. Unfortunately, the basis for this conclusion remains an enigma. Although EGFP labeling was applied in several of the papers that opposed HSC plasticity [139, 140, 145], one of these laboratories introduced the notion that green autofluorescence is commonly present in skeletal muscle samples and therefore the detection of EGFP-positive cells in the heart or skeletal muscle following implantation of EGFP-positive HSCs is by necessity an artifact [146]. This concept was adopted immediately by the International Society of Stem Cell Research [147] and emphasized in original studies and reviews [2, 10]. The rationale for Artifact 5 is incomprehensible. The green autofluorescence observed in histologic sections of skeletal muscle was largely due to the use of an inappropriate fixation protocol for immunolabeling studies. A mixture of paraformaldehyde and glutaraldehyde (Fig. 7A) was used rather than phosphate-buffered formalin (Fig. 7B), which carries minimal background autofluorescence. But most importantly, immunolabeling of glutaraldehyde-fixed tissue never works (Fig. 7C, 7D) because of the high level of cross-linking of proteins generated by glutaraldehyde [148]. Green autofluorescence can be clearly distinguished from EGFP-expressing cells by the use of specific green fluorescence protein antibodies as opposed to direct identification of the EGFP protein by epifluorescence light microscopy (Fig. 7E 7G). Therefore, immunolabeling can be easily optimized so that the emission signal is orders of magnitude greater than background fluorescence, or autofluorescence can be totally excluded by direct labeling of antibodies with quantum dots (Fig. 7H 7K). What remains unclear is the ultimate goal of these unqualified criticisms. They add to the already enormous amount of confusion and uncertainty present in the field of regenerative medicine. The critics of myocardial and skeletal muscle repair by exogenous or endogenous stem cells continue to implement

ENDOGENOUS CARDIAC STEM CELLS IN THE REGENERATION OF ACUTE AND CHRONIC ISCHEMIC MYOCARDIUM

ENDOGENOUS CARDIAC STEM CELLS IN THE REGENERATION OF ACUTE AND CHRONIC ISCHEMIC MYOCARDIUM ENDOGENOUS CARDIAC STEM CELLS IN THE REGENERATION OF ACUTE AND CHRONIC ISCHEMIC MYOCARDIUM Bernardo Nadal-Ginard, M.D., Ph.D. New York Medical College Angioplasty Summit 2004, Seoul 04/29/04 MYOCARDIAL

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature10188 Supplementary Figure 1. Embryonic epicardial genes are down-regulated from midgestation stages and barely detectable post-natally. Real time qrt-pcr revealed a significant down-regulation

More information

Muscle Tissue. General concepts. Classification of muscle. I. Functional classification is based on the type of neural control.

Muscle Tissue. General concepts. Classification of muscle. I. Functional classification is based on the type of neural control. Muscle Tissue LEARNING OBJECTIVES 1. Identify the three types of muscle tissue at the light microscopic level. 2. List and compare the structural and functional features of each of the three muscle fiber

More information

Cardiac Regeneration. Piero Anversa, MD, Annarosa Leri, MD, Jan Kajstura, PHD Valhalla, New York THE HEART AS A POST-MITOTIC ORGAN: THE CONTROVERSY

Cardiac Regeneration. Piero Anversa, MD, Annarosa Leri, MD, Jan Kajstura, PHD Valhalla, New York THE HEART AS A POST-MITOTIC ORGAN: THE CONTROVERSY Journal of the American College of Cardiology Vol. 47, No. 9, 2006 2006 by the American College of Cardiology Foundation ISSN 0735-1097/06/$32.00 Published by Elsevier Inc. doi:10.1016/j.jacc.2006.02.003

More information

The recognition that myocyte mitosis occurs in the fetal,

The recognition that myocyte mitosis occurs in the fetal, Basic Science for Clinicians Life and Death of Cardiac Stem Cells A Paradigm Shift in Cardiac Biology Piero Anversa, MD; Jan Kajstura, PhD; Annarosa Leri, MD; Roberto Bolli, MD The recognition that myocyte

More information

Resident cardiac stem cells: how to find and use them

Resident cardiac stem cells: how to find and use them Resident cardiac stem cells: how to find and use them G. Hasenfuß Cardiology and Pneumology Heart Research Center Göttingen Georg-August-University Göttingen Definition: Stem cell Selfrenewal Stem cell

More information

Medical Biology. Dr. Khalida Ibrahim

Medical Biology. Dr. Khalida Ibrahim Dr. Khalida Ibrahim Medical Biology MUSCLE TISSUE 1. Muscle tissue is characterized by its well-developed properties of contraction. 2. Muscle is responsible for the movements of the body and the various

More information

The New England Journal of Medicine EVIDENCE THAT HUMAN CARDIAC MYOCYTES DIVIDE AFTER MYOCARDIAL INFARCTION

The New England Journal of Medicine EVIDENCE THAT HUMAN CARDIAC MYOCYTES DIVIDE AFTER MYOCARDIAL INFARCTION EVIDENCE THAT HUMAN CARDIAC MYOCYTES DIVIDE AFTER MYOCARDIAL INFARCTION ANTONIO P. BELTRAMI, M.D., KONRAD URBANEK, M.D., JAN KAJSTURA, PH.D., SHAO-MIN YAN, M.D., NICOLETTA FINATO, M.D., ROSSANA BUSSANI,

More information

Supporting Information. Calculation of the relative contributions of myocyte proliferation, stem cell. Supporting Information Fig 1 (page 9)

Supporting Information. Calculation of the relative contributions of myocyte proliferation, stem cell. Supporting Information Fig 1 (page 9) Supporting Information Table of contents Calculation of the relative contributions of myocyte proliferation, stem cell differentiation and cardioprotection (page 2) Supporting Information Fig 1 (page 9)

More information

Mitosis and the Cell Cycle

Mitosis and the Cell Cycle Mitosis and the Cell Cycle Chapter 12 The Cell Cycle: Cell Growth & Cell Division Where it all began You started as a cell smaller than a period at the end of a sentence Getting from there to here Cell

More information

Muscle tissues. Dr. Hersh Abdul Ham-Karim BVM&S, PG Dip, MSc and PhD

Muscle tissues. Dr. Hersh Abdul Ham-Karim BVM&S, PG Dip, MSc and PhD Muscle tissues Dr. Hersh Abdul Ham-Karim BVM&S, PG Dip, MSc and PhD Muscle tissue is a soft tissue that composes muscles in animal bodies, and gives rise to muscles' ability to contract. Muscle tissue

More information

Stretching Cardiac Myocytes: A Finite Element Model of Cardiac Tissue

Stretching Cardiac Myocytes: A Finite Element Model of Cardiac Tissue Megan McCain ES240 FEM Final Project December 19, 2006 Stretching Cardiac Myocytes: A Finite Element Model of Cardiac Tissue Cardiac myocytes are the cells that constitute the working muscle of the heart.

More information

GENERAL HISTOLOGY 4. Muscular Tissue

GENERAL HISTOLOGY 4. Muscular Tissue Biology-232 GENERAL HISTOLOGY 4. Muscular Tissue Dr. Manal Othman Anatomy Department CMMS, AGU Responsible for MOST types of BODY MOVEMENT Made up of groups of elongated MUSCLE cells with contractile filaments

More information

Supplemental Figure 1. Quantification of proliferation in thyroid of WT, Ctns -/- and grafted

Supplemental Figure 1. Quantification of proliferation in thyroid of WT, Ctns -/- and grafted Supplemental Figure 1. Quantification of proliferation in thyroid of WT, Ctns -/- and grafted Ctns -/- mice. Cells immunolabeled for the proliferation marker (Ki-67) were counted in sections (n=3 WT, n=4

More information

Myocardial Regeneration and Stem Cell Repair

Myocardial Regeneration and Stem Cell Repair Myocardial Regeneration and Stem Cell Repair Annarosa Leri, MD, Jan Kajstura, MD, Piero Anversa, MD, and William H. Frishman, MD Abstract: Recent evidence would suggest that the heart is not a terminally

More information

Hematopoiesis. - Process of generation of mature blood cells. - Daily turnover of blood cells (70 kg human)

Hematopoiesis. - Process of generation of mature blood cells. - Daily turnover of blood cells (70 kg human) Hematopoiesis - Process of generation of mature blood cells - Daily turnover of blood cells (70 kg human) 1,000,000,000,000 total cells 200,000,000,000 red blood cells 70,000,000,000 neutrophils Hematopoiesis

More information

The Cell Cycle. Dr. SARRAY Sameh, Ph.D

The Cell Cycle. Dr. SARRAY Sameh, Ph.D The Cell Cycle Dr. SARRAY Sameh, Ph.D Overview When an organism requires additional cells (either for growth or replacement of lost cells), new cells are produced by cell division (mitosis) Somatic cells

More information

Cell cycle and apoptosis

Cell cycle and apoptosis Cell cycle and apoptosis Cell cycle Definition Stages and steps Cell cycle Interphase (G1/G0, S, and G2) Mitosis (prophase, metaphase, anaphase, telophase, karyokinesis, cytokinesis) Control checkpoints

More information

Regenerative Medicine for Cardiomyocytes

Regenerative Medicine for Cardiomyocytes Regenerative Medicine Regenerative Medicine for JMAJ 47(7): 328 332, 2004 Keiichi FUKUDA Assistant Professor, Institute for Advanced Cardiac Therapeutics, Keio University School of Medicine Abstract: Heart

More information

Lecture Overview. CT Framework of the Body. Chapter 4 Muscle & Nervous Tissues Lecture 11. Connective tissue framework of the body

Lecture Overview. CT Framework of the Body. Chapter 4 Muscle & Nervous Tissues Lecture 11. Connective tissue framework of the body Visual Anatomy & Physiology First Edition Martini & Ober Chapter 4 Muscle & Nervous Tissues Lecture 11 Lecture Overview Connective tissue framework of the body Introduction to muscle tissue Classification/characteristics

More information

Skeletal muscle. General features :

Skeletal muscle. General features : Muscular tissues In the first embryonic life the muscular tissues arise from mesoderm, The function of movement in multicellular organisms is usually assumed by specialized cells called muscle fibers which

More information

Cancer Cells. It would take another 20 years and a revolution in the techniques of biological research to answer these questions.

Cancer Cells. It would take another 20 years and a revolution in the techniques of biological research to answer these questions. Cancer Cells Cancer, then, is a disease in which a single normal body cell undergoes a genetic transformation into a cancer cell. This cell and its descendants, proliferating across many years, produce

More information

Supplementary Figure 1. Nature Neuroscience: doi: /nn.4547

Supplementary Figure 1. Nature Neuroscience: doi: /nn.4547 Supplementary Figure 1 Characterization of the Microfetti mouse model. (a) Gating strategy for 8-color flow analysis of peripheral Ly-6C + monocytes from Microfetti mice 5-7 days after TAM treatment. Living

More information

MITOSIS IN DEVELOPING CARDIAC MUSCLE. FRANCIS J. MANASEK. From the Department of Anatomy, Harvard Medical School, Boston, Massachusetts 02115

MITOSIS IN DEVELOPING CARDIAC MUSCLE. FRANCIS J. MANASEK. From the Department of Anatomy, Harvard Medical School, Boston, Massachusetts 02115 Published Online: 1 April, 1968 Supp Info: http://doi.org/10.1083/jcb.37.1.191 Downloaded from jcb.rupress.org on June 30, 2018 MITOSIS IN DEVELOPING CARDIAC MUSCLE FRANCIS J. MANASEK. From the Department

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION DOI: 0.038/ncb33 a b c 0 min 6 min 7 min (fixed) DIC -GFP, CenpF 3 µm Nocodazole Single optical plane -GFP, CenpF Max. intensity projection d µm -GFP, CenpF, -GFP CenpF 3-D rendering e f 0 min 4 min 0

More information

Fig. 1. Simple columnar epithelial cells lining the small intestine.

Fig. 1. Simple columnar epithelial cells lining the small intestine. Mitosis Prelab Reading Fig. 1. Simple columnar epithelial cells lining the small intestine. The tall cells pictured in Fig. 1 form the lining of the small intestine in humans and other animals. These cells

More information

Biology 4A Laboratory MITOSIS Asexual Reproduction OBJECTIVE

Biology 4A Laboratory MITOSIS Asexual Reproduction OBJECTIVE Biology 4A Laboratory MITOSIS Asexual Reproduction OBJECTIVE To study the cell cycle and understand how, when and why cells divide. To study and identify the major stages of cell division. To relate the

More information

Cell Growth and Division *

Cell Growth and Division * OpenStax-CNX module: m46034 1 Cell Growth and Division * OpenStax This work is produced by OpenStax-CNX and licensed under the Creative Commons Attribution License 3.0 By the end of this section, you will

More information

Breaking Up is Hard to Do (At Least in Eukaryotes) Mitosis

Breaking Up is Hard to Do (At Least in Eukaryotes) Mitosis Breaking Up is Hard to Do (At Least in Eukaryotes) Mitosis Chromosomes Chromosomes were first observed by the German embryologist Walther Fleming in 1882. Chromosome number varies among organisms most

More information

Cardiac Stem Cells and Mechanisms of Myocardial Regeneration

Cardiac Stem Cells and Mechanisms of Myocardial Regeneration Physiol Rev 85: 1373 1416, 2005; doi:10.1152/physrev.00013.2005. Cardiac Stem Cells and Mechanisms of Myocardial Regeneration ANNAROSA LERI, JAN KAJSTURA, AND PIERO ANVERSA Cardiovascular Research Institute,

More information

Supplemental Information. Myocardial Polyploidization Creates a Barrier. to Heart Regeneration in Zebrafish

Supplemental Information. Myocardial Polyploidization Creates a Barrier. to Heart Regeneration in Zebrafish Developmental Cell, Volume 44 Supplemental Information Myocardial Polyploidization Creates a Barrier to Heart Regeneration in Zebrafish Juan Manuel González-Rosa, Michka Sharpe, Dorothy Field, Mark H.

More information

meiosis asexual reproduction CHAPTER 9 & 10 The Cell Cycle, Meiosis & Sexual Life Cycles Sexual reproduction mitosis

meiosis asexual reproduction CHAPTER 9 & 10 The Cell Cycle, Meiosis & Sexual Life Cycles Sexual reproduction mitosis meiosis asexual reproduction CHAPTER 9 & 10 The Cell Cycle, Meiosis & Sexual Sexual reproduction Life Cycles mitosis Chromosomes Consists of a long DNA molecule (represents thousands of genes) Also consists

More information

Competency Rubric Bank for the Sciences (CRBS)

Competency Rubric Bank for the Sciences (CRBS) Competency Rubric Bank for the Sciences (CRBS) Content Knowledge 1 Content Knowledge: Accuracy of scientific understanding Higher Order Cognitive Skills (HOCS) 3 Analysis: Clarity of Research Question

More information

Chapter 8 The Cell Cycle

Chapter 8 The Cell Cycle What molecule stores your genetic information or determines everything about you? DNA a nucleic acid How are DNA molecules arranged in the nucleus? As you can see DNA is: Chapter 8 The Cell Cycle 1. Arranged

More information

5.1. KEY CONCEPT Cells have distinct phases of growth, reproduction, and normal functions. 68 Reinforcement Unit 2 Resource Book

5.1. KEY CONCEPT Cells have distinct phases of growth, reproduction, and normal functions. 68 Reinforcement Unit 2 Resource Book 5.1 THE CELL CYCLE KEY CONCEPT Cells have distinct phases of growth, reproduction, and normal functions. Cells have a regular pattern of growth, DNA duplication, and division that is called the cell cycle.

More information

Dako IT S ABOUT TIME. Interpretation Guide. Agilent Pathology Solutions. ALK, ROS1 and RET IQFISH probes (Dako Omnis) MET IQFISH probe (Dako Omnis)

Dako IT S ABOUT TIME. Interpretation Guide. Agilent Pathology Solutions. ALK, ROS1 and RET IQFISH probes (Dako Omnis) MET IQFISH probe (Dako Omnis) INTERPRETATION Dako Agilent Pathology Solutions IQFISH Interpretation Guide ALK, ROS1 and RET IQFISH probes (Dako Omnis) MET IQFISH probe (Dako Omnis) IT S ABOUT TIME For In Vitro Diagnostic Use ALK, ROS1,

More information

DOWNLOAD PDF CARDIAC REMODELING AND CELL DEATH IN HEART FAILURE

DOWNLOAD PDF CARDIAC REMODELING AND CELL DEATH IN HEART FAILURE Chapter 1 : The fibrosis-cell death axis in heart failure Remodeling may be defined as changes in the morphology, structure, and function of the heart related to alterations in loading conditions and/or

More information

chapter 4. The effect of oncogenic HPV on transformation zone epithelium

chapter 4. The effect of oncogenic HPV on transformation zone epithelium chapter 4. The effect of oncogenic HPV on transformation zone epithelium CHAPTER 1 All squamous cervical cancer (and probably all cervical adenocarcinoma) is associated with oncogenic HPV, and the absence

More information

African Trypanosomes

African Trypanosomes African Trypanosomes Giemsa-stained blood smear of African trypanosomes viewed under the 100X objective lens. The block arrows denote trypomastigote forms of the African trypanosomes found within the blood

More information

8 - Muscular System. Introduction Taft College Human Physiology

8 - Muscular System. Introduction Taft College Human Physiology 8 - Muscular System Introduction Taft College Human Physiology Muscular System - Introduction The bones provide the levers and structure of the skeleton but it is the muscles that cause movement. Motion

More information

Cell implantation after myocardial infarction: a 10 years experience from the ICREC laboratory

Cell implantation after myocardial infarction: a 10 years experience from the ICREC laboratory Cell implantation after myocardial infarction: a 10 years experience from the ICREC laboratory BANFF-SCT Joint Scientific Meeting 2017 Barcelona, 29 th March Santi Roura, PhD Grup ICREC IGTP HuGTiP (Badalona)

More information

Breaking Up is Hard to Do (At Least in Eukaryotes) Mitosis

Breaking Up is Hard to Do (At Least in Eukaryotes) Mitosis Breaking Up is Hard to Do (At Least in Eukaryotes) Mitosis Prokaryotes Have a Simpler Cell Cycle Cell division in prokaryotes takes place in two stages, which together make up a simple cell cycle 1. Copy

More information

Exercise 6. Procedure

Exercise 6. Procedure Exercise 6 Procedure Growing of root tips Select a few medium-sized onion bulbs. Carefully remove the dry roots present. Grow root tips by placing the bulbs on glass tubes (of about 3 4 cm. diameter) filled

More information

Myocyte turnover occurs in the human heart, and myocyte

Myocyte turnover occurs in the human heart, and myocyte Intense myocyte formation from cardiac stem cells in human cardiac hypertrophy Konrad Urbanek*, Federico Quaini*, Giordano Tasca, Daniele Torella*, Clotilde Castaldo*, Bernardo Nadal-Ginard*, Annarosa

More information

Cell Division (Mitosis)

Cell Division (Mitosis) Cell Division (Mitosis) Chromosomes The essential part of a chromosome is a single very long strand of DNA. This DNA contains all the genetic information for creating and running the organism. Each chromosome

More information

******************************************************************************************************* MUSCLE CYTOLOGY AND HISTOLOGY

******************************************************************************************************* MUSCLE CYTOLOGY AND HISTOLOGY BIOLOGY 211: HUMAN ANATOMY & PHYSIOLOGY ******************************************************************************************************* MUSCLE CYTOLOGY AND HISTOLOGY *******************************************************************************************************

More information

General Biology. Overview: The Key Roles of Cell Division The continuity of life is based upon the reproduction of cells, or cell division

General Biology. Overview: The Key Roles of Cell Division The continuity of life is based upon the reproduction of cells, or cell division General Biology Course No: BNG2003" Credits: 3.00 " " " 8. The Cell Cycle Prof. Dr. Klaus Heese Overview: The Key Roles of Cell Division The continuity of life is based upon the reproduction of cells,

More information

Mitosis in Onion Root Tip Cells

Mitosis in Onion Root Tip Cells Mitosis in Onion Root Tip Cells A quick overview of cell division The genetic information of plants, animals and other eukaryotic organisms resides in several (or many) individual DNA molecules, or chromosomes.

More information

Mitosis THE CELL CYCLE. In unicellular organisms, division of one cell reproduces the entire organism Multicellular organisms use cell division for..

Mitosis THE CELL CYCLE. In unicellular organisms, division of one cell reproduces the entire organism Multicellular organisms use cell division for.. Mitosis THE CELL CYCLE In unicellular organisms, division of one cell reproduces the entire organism Multicellular organisms use cell division for.. Development from a fertilized cell Growth Repair Cell

More information

Muscle Tissue and Motion

Muscle Tissue and Motion Muscle Tissue and Motion Bởi: OpenStaxCollege Muscle tissue is characterized by properties that allow movement. Muscle cells are excitable; they respond to a stimulus. They are contractile, meaning they

More information

UNC-Duke Biology Course for Residents Fall Cell Cycle Effects of Radiation

UNC-Duke Biology Course for Residents Fall Cell Cycle Effects of Radiation UNC-Duke Biology Course for Residents Fall 2018 1 Cell Cycle: Sequence of changes in a cell starting with the moment the cell is created by cell division, continuing through the doubling of the DNA and

More information

Campbell Biology in Focus (Urry) Chapter 9 The Cell Cycle. 9.1 Multiple-Choice Questions

Campbell Biology in Focus (Urry) Chapter 9 The Cell Cycle. 9.1 Multiple-Choice Questions Campbell Biology in Focus (Urry) Chapter 9 The Cell Cycle 9.1 Multiple-Choice Questions 1) Starting with a fertilized egg (zygote), a series of five cell divisions would produce an early embryo with how

More information

Cell Division Questions. Mitosis and Meiosis

Cell Division Questions. Mitosis and Meiosis Cell Division Questions Mitosis and Meiosis 1 10 Do not write outside the box 5 Figure 3 shows a pair of chromosomes at the start of meiosis. The letters represent alleles. Figure 3 E E e e F F f f 5 (a)

More information

1. Cardiomyocytes and nonmyocyte. 2. Extracellular Matrix 3. Vessels שאלה 1. Pathobiology of Heart Failure Molecular and Cellular Mechanism

1. Cardiomyocytes and nonmyocyte. 2. Extracellular Matrix 3. Vessels שאלה 1. Pathobiology of Heart Failure Molecular and Cellular Mechanism Pathobiology of Heart Failure Molecular and Cellular Mechanism Jonathan Leor Neufeld Cardiac Research Institute Tel-Aviv University Sheba Medical Center, Tel-Hashomer שאלה 1 התא הנפוץ ביותר (75%~) בלב

More information

Data Fact Sheet. Congestive Heart Failure in the United States: A New Epidemic

Data Fact Sheet. Congestive Heart Failure in the United States: A New Epidemic National Heart, Lung, and Blood Institute Data Fact Sheet Congestive Heart Failure National Heart, Lung, and Blood Institute National Institutes of Health Data Fact Sheet Congestive Heart Failure in the

More information

The pathogenesis of nervous distemper

The pathogenesis of nervous distemper Veterinary Sciences Tomorrow - 2004 The pathogenesis of nervous distemper Marc Vandevelde Canine distemper is a highly contagious viral disease of dogs and of all animals in the Canidae, Mustellidae and

More information

Biology is the only subject in which multiplication is the same thing as division

Biology is the only subject in which multiplication is the same thing as division Biology is the only subject in which multiplication is the same thing as division 2007-2008 The Cell Cycle: Cell Growth, Cell Division Ch. 10 Where it all began You started as a cell smaller than a period

More information

Essential Questions. Why are cells relatively small? What are the primary stages of the cell cycle? What are the stages of interphase?

Essential Questions. Why are cells relatively small? What are the primary stages of the cell cycle? What are the stages of interphase? Essential Questions Why are cells relatively small? What are the primary stages of the cell cycle? What are the stages of interphase? Cellular Growth Vocabulary Review selective permeability New cell cycle

More information

International Graduate Research Programme in Cardiovascular Science

International Graduate Research Programme in Cardiovascular Science 1 International Graduate Research Programme in Cardiovascular Science This work has been supported by the European Community s Sixth Framework Programme under grant agreement n LSHM-CT-2005-01883 EUGeneHeart.

More information

The Cell Cycle and How Cells Divide

The Cell Cycle and How Cells Divide The Cell Cycle and How Cells Divide 1 Phases of the Cell Cycle The cell cycle consists of Interphase normal cell activity The mitotic phase cell divsion INTERPHASE Growth G 1 (DNA synthesis) Growth G 2

More information

Cellular Reproduction

Cellular Reproduction 9 Cellular Reproduction section 1 Cellular Growth Before You Read Think about the life cycle of a human. On the lines below, write some of the stages that occur in the life cycle of a human. In this section,

More information

Genetics and Cellular Function

Genetics and Cellular Function Genetics and Cellular Function DNA replication and the cell cycle Mitosis Mitosis Mitosis: division of cells that results in daughter cells with the same the genetic information that the original cell

More information

KEY CONCEPT Cells have distinct phases of growth, reproduction, and normal functions.

KEY CONCEPT Cells have distinct phases of growth, reproduction, and normal functions. 5.1 The Cell Cycle KEY CONCEPT Cells have distinct phases of growth, reproduction, and normal functions. Objective: Cells have distinct phases of growth, reproduction and normal functions. APK: Why do

More information

Biology is the only subject in which multiplication is the same thing as division. AP Biology

Biology is the only subject in which multiplication is the same thing as division. AP Biology Biology is the only subject in which multiplication is the same thing as division Chapter 12. The Cell Cycle: Cell Growth, Cell Division Where it all began You started as a cell smaller than a period at

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION b 350 300 250 200 150 100 50 0 E0 E10 E50 E0 E10 E50 E0 E10 E50 E0 E10 E50 Number of organoids per well 350 300 250 200 150 100 50 0 R0 R50 R100 R500 1st 2nd 3rd Noggin 100 ng/ml Noggin 10 ng/ml Noggin

More information

Cellular Reproduction

Cellular Reproduction Section 1: Cellular Growth Section 2: Mitosis and Cytokinesis Section 3: Cell Cycle Regulation Click on a lesson name to select. Section 1 Cellular Growth Ratio of Surface Area to Volume Section 1 Cellular

More information

Ploidy and Human Cell Types. Cell Cycle and Mitosis. DNA and Chromosomes. Where It All Began 11/19/2014. Chapter 12 Pg

Ploidy and Human Cell Types. Cell Cycle and Mitosis. DNA and Chromosomes. Where It All Began 11/19/2014. Chapter 12 Pg Ploidy and Human Cell Types Cell Cycle and Mitosis Chapter 12 Pg. 228 245 Cell Types Somatic cells (body cells) have 46 chromosomes, which is the diploid chromosome number. A diploid cell is a cell with

More information

Microanatomy of Muscles. Anatomy & Physiology Class

Microanatomy of Muscles. Anatomy & Physiology Class Microanatomy of Muscles Anatomy & Physiology Class Three Main Muscle Types Objectives: By the end of this presentation you will have the information to: 1. 2. 3. 4. 5. 6. Describe the 3 main types of muscles.

More information

General Biology. Overview: The Key Roles of Cell Division. Unicellular organisms

General Biology. Overview: The Key Roles of Cell Division. Unicellular organisms General Biology Course No: BNG2003 Credits: 3.00 8. The Cell Cycle Prof. Dr. Klaus Heese Overview: The Key Roles of Cell Division The continuity of life is based upon the reproduction of cells, or cell

More information

Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future

Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future Cell Therapy 2014 Las Vegas, NV, USA Sulaiman Al-Hashmi, PhD Sultan Qaboos University Oman What are MSCs? Stem

More information

Cell Cycle and Mitosis

Cell Cycle and Mitosis Cell Cycle and Mitosis Name Period A# THE CELL CYCLE The cell cycle, or cell-division cycle, is the series of events that take place in a eukaryotic cell between its formation and the moment it replicates

More information

Regulation of the Cell Cycle

Regulation of the Cell Cycle Regulation of the Cell Cycle 21 I. OVERVIEW Quiescent differentiated cell / can be induced to re-enter the active cell cycle. urvival Cell division Apoptosis 1 Daughter cells Apoptic cell enescent cell

More information

Mitosis Notes AP Biology Mrs. Laux

Mitosis Notes AP Biology Mrs. Laux I. Cell Cycle-includes interphase and mitosis (IPPMAT) A. Interphase 1. accounts for 90% of the cycle 2. cell grows and copies its chromosomes in preparation for cell division 3. produces proteins and

More information

LYMPH GLAND. By : Group 1

LYMPH GLAND. By : Group 1 LYMPH GLAND By : Group 1 ANATOMY LYMPH NODE Lymphatic Organs Red bone marrow Thymus gland Lymph nodes Lymph nodules Spleen Primary organs Secondary organs Lymph Nodes Firm, smooth-surfaced, bean-shaped

More information

Regulators of Cell Cycle Progression

Regulators of Cell Cycle Progression Regulators of Cell Cycle Progression Studies of Cdk s and cyclins in genetically modified mice reveal a high level of plasticity, allowing different cyclins and Cdk s to compensate for the loss of one

More information

Cell Growth and Division

Cell Growth and Division Name Class Date 10 Cell Growth and Division Big idea Growth, Development, and Reproduction Q: How does a cell produce a new cell? WHAT I KNOW WHAT I LEARNED 10.1 Why do cells divide? 10.2 How do cells

More information

Chapter 12. living /non-living? growth repair renew. Reproduction. Reproduction. living /non-living. fertilized egg (zygote) next chapter

Chapter 12. living /non-living? growth repair renew. Reproduction. Reproduction. living /non-living. fertilized egg (zygote) next chapter Chapter 12 How cells divide Reproduction living /non-living? growth repair renew based on cell division first mitosis - distributes identical sets of chromosomes cell cycle (life) Cell Division in Bacteria

More information

NUCLEAR ABNORMALITIES RESULTING FROM INHIBITION OF MITOSIS BY COLCHICINE AND OTHER SUBSTANCES '

NUCLEAR ABNORMALITIES RESULTING FROM INHIBITION OF MITOSIS BY COLCHICINE AND OTHER SUBSTANCES ' NUCLEAR ABNORMALITIES RESULTING FROM INHIBITION OF MITOSIS BY COLCHICINE AND OTHER SUBSTANCES ' AUSTIN M. BRUES AND ELIZABETH B. JACKSON (From the Medicul Lnborutorif~ of the Collis P. Iiz4ntingtolz M~moricil

More information

Cellular Reproduction

Cellular Reproduction Cellular Reproduction How do we know that cells divide to form other cells? Growth Chapter 9 Cuts or wounds heal New blood is produced Hair and fingernails grow back Do cells grow? 1 Hollywood Limits to

More information

Biology is the only subject in which multiplication is the same thing as division

Biology is the only subject in which multiplication is the same thing as division The Cell Cycle Biology is the only subject in which multiplication is the same thing as division Why do cells divide? For reproduction asexual reproduction For growth one-celled organisms from fertilized

More information

Cell Cycle. Trends in Cell Biology

Cell Cycle. Trends in Cell Biology Cell Cycle Trends in Cell Biology Cell Cycle The orderly sequence of events by which a cell duplicates its contents and divides into two Daughter Cells Activities of a cell from one cell division to the

More information

STEM CELL RESEARCH: MEDICAL PROGRESS WITH RESPONSIBILITY

STEM CELL RESEARCH: MEDICAL PROGRESS WITH RESPONSIBILITY STEM CELL RESEARCH: MEDICAL PROGRESS WITH RESPONSIBILITY A REPORT FROM THE CHIEF MEDICAL OFFICER S EXPERT GROUP REVIEWING THE POTENTIAL OF DEVELOPMENTS IN STEM CELL RESEARCH AND CELL NUCLEAR REPLACEMENT

More information

CELL DIVISION MITOSIS ANSWERS TO PAST PAPER QUESTIONS

CELL DIVISION MITOSIS ANSWERS TO PAST PAPER QUESTIONS CELL DIVISION MITOSIS ANSWERS TO PAST PAPER QUESTIONS Question Marking details 1 (a) (i) Telophase (1) Accept cytokinesis Metaphase (1) Any reference to meiosis penalise 1 mark (ii) Chromosomes Accept

More information

Copyright The McGraw-Hill Companies, Inc. Permission required for reproduction or display. Chapter 9 MITOSIS

Copyright The McGraw-Hill Companies, Inc. Permission required for reproduction or display. Chapter 9 MITOSIS Copyright The McGraw-Hill Companies, Inc. Permission required for reproduction or display. Chapter 9 MITOSIS Copyright The McGraw-Hill Companies, Inc. Permission required for reproduction or display. 9.1

More information

Integrated Muscle. Red: important. Black: in male female slides. Gray: notes extra. Editing File

Integrated Muscle. Red: important. Black: in male female slides. Gray: notes extra. Editing File Integrated Muscle Red: important. Black: in male female slides. Gray: notes extra. Editing File OBJECTIVES Identify and describe the histological structure of the three types of muscle cells and list the

More information

Part II The Cell Cell Division, Chapter 2 Outline of class notes

Part II The Cell Cell Division, Chapter 2 Outline of class notes Part II The Cell Cell Division, Chapter 2 Outline of class notes 1 Cellular Division Overview Types of Cell Division Chromosomal Number The Cell Cycle Mitoses Cancer Cells In Vitro Fertilization Infertility

More information

CELL CYCLE INTRODUCTION PART I ANIMAL CELL CYCLE INTERPHASE

CELL CYCLE INTRODUCTION PART I ANIMAL CELL CYCLE INTERPHASE CELL CYCLE INTRODUCTION The nuclei in cells of eukaryotic organisms contain chromosomes with clusters of genes, discrete units of hereditary information consisting of double-stranded DNA. Structural proteins

More information

Table of Contents. 1. Overview. 2. Interpretation Guide. 3. Staining Gallery Cases Negative for CINtec PLUS

Table of Contents. 1. Overview. 2. Interpretation Guide. 3. Staining Gallery Cases Negative for CINtec PLUS Staining Atlas Table of Contents 1. Overview 1.1 Introduction 1.2 Role of p16 INK4a 1.3 Role of Ki-67 1.4 Molecular Pathogenesis 1.5 p16 INK4a Expression in Cervical Dysplasia 1.6 The Concept of CINtec

More information

Histology of the myocardium and blood vessels. Prof. Abdulameer Al-Nuaimi

Histology of the myocardium and blood vessels. Prof. Abdulameer Al-Nuaimi Histology of the myocardium and blood vessels Prof. Abdulameer Al-Nuaimi E-mail: a.al-nuaimi@sheffield.ac.uk E-mail: abdulameerh@yahoo.com Histology of blood vessels The walls of arteries and veins are

More information

Outline Interphase Mitotic Stage Cell Cycle Control Apoptosis Mitosis Mitosis in Animal Cells Cytokinesis Cancer Prokaryotic Cell Division

Outline Interphase Mitotic Stage Cell Cycle Control Apoptosis Mitosis Mitosis in Animal Cells Cytokinesis Cancer Prokaryotic Cell Division The Cell Cycle and Cellular Reproduction Chapter 9 Outline Interphase Mitotic Stage Cell Cycle Control Apoptosis Mitosis Mitosis in Animal Cells Cytokinesis Cancer Prokaryotic Cell Division 1 2 Interphase

More information

Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy

Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy Massimiliano Gnecchi, Zhiping Zhang, Aiguo Ni, Victor J. Dzau Circulation Research 2008 Nov 21;103(11):1204-19 Introduction(1) After AMI all

More information

Chapter 2. Mitosis and Meiosis

Chapter 2. Mitosis and Meiosis Chapter 2. Mitosis and Meiosis Chromosome Theory of Heredity What structures within cells correspond to genes? The development of genetics took a major step forward by accepting the notion that the genes

More information

SUPPLEMENTAL INFORMATION FOR. PAX7 expression defines germline stem cells in the adult testis

SUPPLEMENTAL INFORMATION FOR. PAX7 expression defines germline stem cells in the adult testis SUPPLEMENTAL INFORMATION FOR PAX7 expression defines germline stem cells in the adult testis Gina M. Aloisio, Yuji Nakada, Hatice D. Saatcioglu, Christopher G. Peña, Michael D. Baker, Edward D. Tarnawa,

More information

Tissue renewal and Repair. Nisamanee Charoenchon, PhD Department of Pathobiology, Faculty of Science

Tissue renewal and Repair. Nisamanee Charoenchon, PhD   Department of Pathobiology, Faculty of Science Tissue renewal and Repair Nisamanee Charoenchon, PhD Email: nisamanee.cha@mahidol.ac.th Department of Pathobiology, Faculty of Science Topic Objectives 1. Describe processes of tissue repair, regeneration

More information

Extra notes for lab- 1 histology. Slide 1 : cross section in the elastic artery ( aortic arch, ascending aorta, descending aorta )

Extra notes for lab- 1 histology. Slide 1 : cross section in the elastic artery ( aortic arch, ascending aorta, descending aorta ) Extra notes for lab- 1 histology Slide 1 : cross section in the elastic artery ( aortic arch, ascending aorta, descending aorta ) - twin of ascending aorta is the pulmonary trunk. Ascending aorta represents

More information

T R L J. Version 2, 2018 NAME: OPTION GROUP: CELL DIVISION MITOSIS WORKBOOK

T R L J. Version 2, 2018 NAME: OPTION GROUP: CELL DIVISION MITOSIS WORKBOOK NAME: OPTION GROUP: CELL DIVISION MITOSIS WORKBOOK 1 STUDY CHECKLIST AND ASSESSMENT OBJECTIVES Instructions Regular revision throughout the year is essential. It s vital you keep a track of what you understand

More information

Cell Death & Renewal (part 2)

Cell Death & Renewal (part 2) 17 Cell Death & Renewal (part 2) Programmed Cell Death A major signaling pathway that promotes cell survival is initiated by the enzyme PI 3-kinase, which phosphorylates PIP2 to form PIP3, which activates

More information

MITOSIS IN ONION ROOTLET CELLS

MITOSIS IN ONION ROOTLET CELLS Lesson 6: CELL CYCLE, MITOSIS Name: Group: MITOSIS IN ONION ROOTLET CELLS Permanent slide: onion rootlet stained with acetorcein The particular mitotic phases are visible in the onion rootlet cells. Chromosomes

More information

Lab Animal Tissue. LEARNING OBJECTIVES: To understand the relationship between the structure and function of different animal tissues

Lab Animal Tissue. LEARNING OBJECTIVES: To understand the relationship between the structure and function of different animal tissues Name: Bio A.P. PURPOSE: HYPOTHESIS: NONE Lab Animal Tissue BACKGROUND: In animals, groups of closely related cells specialized to perform the same function are called tissues. There are four general classes

More information

Muscle Tissue. Muscle Development and Repair. Development: fusion of myoblasts. Repair: Satellite cells (S) 3 Types of Muscle

Muscle Tissue. Muscle Development and Repair. Development: fusion of myoblasts. Repair: Satellite cells (S) 3 Types of Muscle ANNOUNCEMENTS Review Session Every Friday at 12:20 Muscle Tissue 3 Types of Muscle Function: Force generation Lab Practical Coming up! October 26 th, 27 th Muscle Tissue Striated Nonstriated Skeletal Smooth

More information